• No results found

Zr-89-pembrolizumab biodistribution is influenced by PD-1-mediated uptake in lymphoid organs

N/A
N/A
Protected

Academic year: 2021

Share "Zr-89-pembrolizumab biodistribution is influenced by PD-1-mediated uptake in lymphoid organs"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Zr-89-pembrolizumab biodistribution is influenced by PD-1-mediated uptake in lymphoid

organs

van der Veen, Elly L.; Giesen, Danique; Pot-de Jong, Linda; Jorritsma-Smit, Annelies; De

Vries, Elisabeth G. E.; Lub-de Hooge, Marjolijn N.

Published in:

Journal for immunotherapy of cancer DOI:

10.1136/jitc-2020-000938

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

van der Veen, E. L., Giesen, D., Pot-de Jong, L., Jorritsma-Smit, A., De Vries, E. G. E., & Lub-de Hooge, M. N. (2020). Zr-89-pembrolizumab biodistribution is influenced by PD-1-mediated uptake in lymphoid organs. Journal for immunotherapy of cancer, 8(2), [000938]. https://doi.org/10.1136/jitc-2020-000938

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

89

Zr- pembrolizumab biodistribution is

influenced by PD-1- mediated uptake in

lymphoid organs

Elly L van der Veen ,1 Danique Giesen,1 Linda Pot- de Jong,1

Annelies Jorritsma- Smit,2 Elisabeth G E De Vries,1 Marjolijn N Lub- de Hooge 2,3

To cite: van der Veen EL,

Giesen D, Pot- de Jong L,

et al. 89Zr- pembrolizumab

biodistribution is influenced by PD-1- mediated uptake in lymphoid organs. Journal for ImmunoTherapy of Cancer

2020;8:e000938. doi:10.1136/

jitc-2020-000938

►Additional material is

published online only. To view please visit the journal online (http:// dx. doi. org/ 10. 1136/ jitc- 2020- 000938).

Accepted 11 August 2020

1Department of Medical

Oncology, UMCG, Groningen, Groningen, Netherlands

2Department of Clinical

Pharmacy and Pharmacology, UMCG, Groningen, Groningen, Netherlands

3Department of Nuclear

Medicine and Molecular Imaging, UMCG, Groningen, Groningen, Netherlands

Correspondence to

Dr Marjolijn N Lub- de Hooge; m. n. de. hooge@ umcg. nl © Author(s) (or their employer(s)) 2020. Re- use permitted under CC BY- NC. No commercial re- use. See rights and permissions. Published by BMJ.

ABSTRACT

Background To better predict response to immune

checkpoint therapy and toxicity in healthy tissues, insight in the in vivo behavior of immune checkpoint targeting monoclonal antibodies is essential. Therefore, we aimed to study in vivo pharmacokinetics and whole- body distribution of zirconium-89 (89Zr) labeled programmed

cell death protein-1 (PD-1) targeting pembrolizumab with positron- emission tomography (PET) in humanized mice.

Methods Humanized (huNOG) and non- humanized NOG

mice were xenografted with human A375M melanoma cells. PET imaging was performed on day 7 post 89Zr-

pembrolizumab (10 µg, 2.5 MBq) administration, followed by ex vivo biodistribution studies. Other huNOG mice bearing A375M tumors received a co- injection of excess (90 µg) unlabeled pembrolizumab or 89Zr- IgG

4 control

(10 µg, 2.5 MBq). Tumor and spleen tissue were studied with autoradiography and immunohistochemically including PD-1.

Results PET imaging and biodistribution studies showed

high 89Zr- pembrolizumab uptake in tissues containing

human immune cells, including spleen, lymph nodes and bone marrow. Tumor uptake of 89Zr- pembrolizumab was

lower than uptake in lymphoid tissues, but higher than uptake in other organs. High uptake in lymphoid tissues could be reduced by excess unlabeled pembrolizumab. Tracer activity in blood pool was increased by addition of unlabeled pembrolizumab, but tumor uptake was not affected. Autoradiography supported PET findings and immunohistochemical staining on spleen and lymph node tissue showed PD-1 positive cells, whereas tumor tissue was PD-1 negative.

Conclusion 89Zr- pembrolizumab whole- body

biodistribution showed high PD-1- mediated uptake in lymphoid tissues, such as spleen, lymph nodes and bone marrow, and modest tumor uptake. Our data may enable evaluation of 89Zr- pembrolizumab whole- body distribution

in patients.

BACKGROUND

Immune checkpoint inhibitors targeting the programmed cell death protein-1 (PD-1/ programmed death ligand-1 (PD- L1) pathway are showing impressive antitumor effects. However, not all patients respond and serious immune- related toxicity has been reported.1 This has raised interest in

better understanding the behavior of these drugs in the human body. PD- L1 and PD-1 are expressed by a broad range of immune cells, including T- cells, B- cells, natural killer (NK) cells, monocytes and dendritic cells. PD- L1 can be highly expressed by tumor cells, whereas PD-1 expression is most prominent in T- cells and lower in other immune cells.2

Biodistribution of PD-1 and PD- L1 targeting drugs will likely be influenced by the dynamic expression patterns of these targets.

Molecular imaging has proven to be an useful tool for studying drug biodistribu-tion.3 4 In table 1, we summarized preclinical

imaging studies that investigated biodistribu-tion of radiolabeled molecules targeting PD-1 and PD- L1.5–28 Most studies that we reviewed

focused on tracer distribution in the tumor and its microenvironment, without consid-ering PD-1 and PD- L1 expression in healthy immune tissues. Studies that do report on tracer uptake in lymphoid tissues are scarce and results are often limited to the spleen. Furthermore, most tracers targeting human PD-1/PD- L1 are not cross- reactive with murine proteins and relevant mouse models reconsti-tuted with (parts of) a human immune system are rarely used. A limited number of studies used NOD scid gamma (NSG) mice engrafted with human peripheral blood mononuclear cells (hNSG model).23–25 27 The hNSG model

has a high level of functional T- cells, however, it is also characterized by aberrant distribution of immune cells to murine immune tissues and other cell lineages remain underdevel-oped.29 Humanized mice that are engrafted with human CD34 + hematopoietic stem cells (HSCs) establish an immune- competent model with a broader set of developed human immune cells present and might therefore be a better surrogate for the human immune environment.

To gain more insight in the in vivo behavior of a human PD-1 targeting monoclonal

copyright.

on November 18, 2020 at University of Groningen. Protected by

(3)

Table 1

Pr

eclinical imaging studies tar

geting

L1 and PD-1, using radiolabeled monoclonalantibody or small pr

oteins

Type of Imaging

Tracer

Origin and reactivity

Cr oss reactivity Animal model Tumor model Tracer dose

Imaging / biodistribution time point

Tumor uptake

Uptake lymphoid tissue

Ref L1 - antibodies SPECT/CT 111 L1.3.1 antibody

Murine anti- human

No

Balb/c nude mice 6 to 8 weeks old Immune deficient

Human br

east

cancer cell lines

1.5

µg (15.5 MBq) and

1.0

µg (10.0 MBq)

Imaging and ex vivo biodistribution at 24, 72 and 168

hours pi

32.8 (±6.8) %ID/g and 6.2 (±1.0) %ID/g at 168

hours pi for MB-231 and MCF-7 tumors r espectively L1 detection at dif fer ent expr ession levels in Br -3, SUM149 and BT474 tumors No ( 5 ) SPECT 111 DTP L1 antibody

Hamster anti- mouse

No

neu

- N transgenic mice

8 to 12 weeks old Immune competent NT2.5 (mouse mammary tumor) 7.4 MBq for imaging and 8.4

µg (0.93 MBq) for

biodistribution

Imaging on 1, 24, and 72 days pi and ex vivo biodistribution at 1, 24, 72, and 144

hours pi

Tumor uptake of 21.1 (±11.2) %ID/g at 144

hours

pi

Yes, spleen (63.5%±25.4 %ID/g) and thymus (16.8%±16.2 %ID/g) at 144

hours pi

Spleen uptake was blocked by coinjection of unlabeled antibody

( 6 ) SPECT 111 L1 antibody

Humanized anti- human

Cr

reactive

with mouse

NSG mice 6 to 8 weeks old Immune deficient

Human cell lines

100

µg (14.8 MBq) for

imaging and 8.5

µg (1.48

MBq) for biodistribution

Imaging and ex vivo biodistribution at 24, 48, 72, 96 and 120

hours pi

8.9 (±0.26) %ID/g at 72 hours pi for MDA- MB-231 tumors and 7.46 (±0.12) at 144

hours pi for H2444 tumors Detection of L1 at dif fer ent expr ession levels Yes, spleen (23.5±8.2) at 48 hours pi

Spleen uptake was blocked by

injection of unlabeled antibody ( 7 ) PET 64 L1 antibody

Humanized anti- human

Cr

reactive

with mouse

NSG mice 6 to 8 weeks old Immune deficient

Human cell lines

16.7 MBq (40

µg) for

imaging and 1.48 MBq (10

µg) for biodistribution

Imaging on 2, 24 and 48 hours pi and ex vivo biodistribution at 24 and 48 hours

pi

40.6 (±6.9) %ID/g, 17.2 (±2.1) %ID/g and 9.4 (±2.3) %ID/g at 48

hours pi for L1 positive CHO, MB-231

and SUM149 tumors respectively

High spleen uptake (~45 %ID/g) at 24

hours

pi after blocking with unlabeled antibody

(

8

)

Balb/c mice 4 to 6 weeks old Immune competent 4T1 (mouse mammary car

cinoma)

17.0 (±4.3) %ID/g at 48 hours pi for 4T1 tumors No high uptake observed in spleen (±12 %ID/g) and BA

T SPECT 111 DTP L1 antibody Rat mouse No

C57BL/6 mice 6 to 8 weeks old Immune competent B16F10 (murine melanoma) 15–16 MBq (60 µg) for imaging and 0.37 MBq (0.13 mg/kg) Imaging on 1, 24 and 72 hours pi and biodistribution at 1, 24, 72 and 96 hours pi

6.6 (±3.1) %ID/g at 24 hours pi for B16F10 tumors

Yes, spleen (47%±9.5 %ID/g) at 24

hours pi

and BA

T

Spleen uptake was blocked by coinjection of unlabeled antibody

( 9 ) PET 89Zr - L1 antibody Rat mouse No

C57BL/6 mice 6 to 8 weeks old Immune competent MEER (murine tonsil epithelium) or B16F10 (murine melanoma)

3.7 MBq (50

µg)

Imaging and ex vivo biodistribution at 48 and 96 hours

pi

Higher uptake in irradiated (20.1%±2.6 %ID/g) vs

irradiated

(11.1%±1.9

%ID/g) MEER tumors Higher uptake in irradiated (28.0%±4.9 %ID/g) vs

irradiated

(14.4%±1.4

%ID/g) B16F10 tumors

Yes, spleen (60% to 120%ID/g) and thymus (25% to 35%ID/g) Spleen uptake was blocked by pr

injection of unlabeled antibody ( 10 ) PET 89Zr - C4

(recombinant IgG1 antibody)

Engineer ed anti-human Cr reactive mouse

Nu/nu mice 3 to 5 weeks old Immune deficient H1975 and A549 (human NSCLC), PC3 (human prostatic small cell car

cinoma)

11.1 MBq for imaging and 1.85 MBq for ex vivo biodistribution Imaging and ex vivo biodistribution at 8, 24, 48, 72, 120

hours pi

~9 %ID/g~5 %ID/g and ~7 %ID/g at 48

hours pi

for H1975, A459 and PC3 tumors r

espectively

~13%

ID/g at 48

hours pi

for B16F16 tumors ~5 %ID/g tumor uptake at 48 hours pi in PDX model Detection of

induced

changes in

L1

expr

ession

Yes, spleen uptake of ~7 %ID/g and ~6 %ID/g at 48

hours pi in nu/nu and C57BL/6

mice r

espectively

Incr

eased uptake in the spleens of nu/nu

mice tr

eated with paclitaxel and spleens of

C57BL/6 mice tr

eated with doxorubicin

(

11

)

C57BL/6 mice 3 to 5 weeks old Immune deficient B16F10 (mouse melanoma)

Not r

eported

PDX model of EGFR mutant (L858R) NSCLC

Continued

copyright.

on November 18, 2020 at University of Groningen. Protected by

(4)

Type of Imaging

Tracer

Origin and reactivity

Cr oss reactivity Animal model Tumor model Tracer dose

Imaging / biodistribution time point

Tumor uptake

Uptake lymphoid tissue

Ref SPECT/CT 111 L1 Rat murine No

Balb/c and C57BL/6 6 to 8 weeks old Immune competent

Murine cell lines

19.7 (±1.2) MBq (30

µg)

Imaging and ex vivo biodistribution at 72

hours

pi

~14.53 (±5.49) %ID/g,~16.29 (±5.57) %ID/g,~11.06 (±6.54) %ID/g,~14.94 (±4.01) %ID/g,~6.16 (±2.94) %ID/g, for Renca, 4T1, CT26, B16F1 and LLC1 respectively

Yes, spleen varying fr

om 13.09 %ID/g to

40.30 %ID/g. Thymus varying fr

om 6.09 %ID/g to 10.26 %ID/g ( 12 ) 111 L1

Murine anti- human

No humanized and humanized NSG mice MB-231 (human br east car cinoma) 11.9±1.6 MBq (1 µg) 111 L1 Or 11.5±0.4 MBq (2.8 µg) 111 contr ol mIgG1 LPS tr eatment 1 day befor e tracer injection

Imaging and ex vivo biodistribution at 72

hours

pi

~40 %ID/g for non- humanized mice and ~60 %ID/g for humanized mice at 72

hours pi

~35 %ID/g for humanized mice after LPS tr

eatment at 72 hours pi ~8 %ID/g for 111 contr ol

mIgG1 in both non- humanized and humanized mice at 72

hours pi

Yes, spleen uptake ~20%

ID/g for

non-humanize mice and ~25%

ID/g for

humanized mice ~80 %ID/g for humanized mice after LPS treatment at 72

hours pi ~10 %ID/g for 111 contr ol mIgG1 (both gr oups 111 L1 Rat murine No

Balb/c and C57BL/6 6 to 8 weeks old Immune competent

Murine cell lines

Irradiation followed on day one by injection of 23.8±1.7

MBq

(30

µg)

Imaging and ex vivo biodistribution at 24

hours

pi

Higher uptake in irradiated (26.3%±2.0 %ID/g) vs

irradiated

(17.1%±3.1

%ID/g) CT26 tumors Higher uptake in irradiated (15.7%±1.8 %ID/g) vs

irradiated (12.3%±1.7% ID/g) LLC1 tumors No dif fer ence uptake in

irradiated (14.9%±6.8 %ID/g) vs

irradiated

(16.7%±3.5%

ID/g)

for

B16F1 tumors

Spleen uptake ~14% to 17%ID/g for all models Higher uptake in lymph nodes of irradiated tumor models vs

irradiated tumor models L1 - small molecules PET 64 WL12 L1 binding peptide) Engineer ed anti-human No

NSG mice 6 to 8 weeks old Immune deficient

High L1-expr essing CHO cell line

5.6 MBq for imaging and 1.5 MBq for ex vivo biodistribution Imaging and ex vivo biodistribution at 10

min, 0.5, 1 and 2 hour pi 14.9 (±0.8) at 1 hour pi in expr essing CHO tumors No ( 13 ) PET 18 NOT A-Z L1_ (anti L1

small molecule, affibody)

Engineer

ed

anti-human af

fibody

No

SCID beige mice 6 to 8 weeks old Immune deficient

IMVI

(human

melanoma) and SUDHL6 (human B- cell

lymphoma)

0.2 to 0.6 MBq Dynamic PET scan during 90 min 2.56 (±0.33) %ID/g at 90 min pi for LOX tumors

No ( 14 ) SPECT 99m L1 nanobodies Engineer ed mouse nanobodies Cr reactive human C57BL/6 mice (WT) vs CD8 depleted L1

KO mice 6 weeks old Immune competent TC-1 (mouse lung epithelial), WT TC-1

L1+ vs CRISPR/Cas9- modified TC-1 L1 KO 45 to 155 MBq (10 µg) nanobody Imaging 1 hour pi and

ex vivo biodistribution 80 min

pi

1.7 (±0.1) %ID/g for WT and 1.1 (±0.3) %ID/g for KO at 80

min pi

Yes, spleen 11.4 (±1.4) %ID/g for WT and 1.6±0.2%

ID/g for KO at 80

min pi

Lymph node uptake 3.5 (±0.8) %ID/g for WT and 0.4 (±0.1) %ID/g for KO at 80

min pi ( 15 ) PET 64 PD-1

ectodomain targeting

L1 Engineer ed anti-human Not specified

NSG mice Immune deficient CT26 (mouse colon cancer)

L1 (+) or L1(-) 8.5 MB (25 µg) Imaging at 1, 2, 4, and 24 hours. Ex vivo

biodistribution at 1 and 24 hours

~3 %ID/g for

L1 (+)

and ~1.8 %ID/g for

L1 (-) at

24

hours

pi

Yes, spleen ~5 %ID/g at 24

hours pi ( 16 ) Table 1 Continued Continued copyright.

on November 18, 2020 at University of Groningen. Protected by

(5)

Type of Imaging

Tracer

Origin and reactivity

Cr oss reactivity Animal model Tumor model Tracer dose

Imaging / biodistribution time point

Tumor uptake

Uptake lymphoid tissue

Ref PET 18 BMS-986192 L1 small molecule) Engineer ed anti-human Af finity for

human & cynomolgus

L1,

no

binding to murine

L1)

Immune deficient mice

Human L2987 L1+) and HT -29 L1-) 5.6 MBq, block to 3 mg/kg Dynamic PET scan during 120

min

2.41 (±0.29) %ID/g for

L1 +and 0.82 (±0.11)

%ID/g for

L1-, 0.79

(±0.12) %ID/g after blocking in

L+

Yes, spleen uptake (no clear numbers)

( 17 ) Cynomolgus monkeys – 55.5 MBq

Dynamic PET scan during 150

min

Yes, spleen:muscle 12:1, after blocking spleen:muscle1.24:1

PET 64 PD-1 ectodomains (DOT A-/ NOT HAC, aglycosylated DOT A-/NOT A-HACA) Engineer ed anti-human Not specified

NSG mice 6 to 8 weeks old Immune deficient CT26 (mouse colon cancer)

L1(+)

or

L1(-)

0.7–3.7 MBq (10 to 15 µg) Imaging and ex vivo biodistribution at 1

hour pi

1.8 (±0.2) %ID/g for PD- L1(+) and 0.9 (±0.7) %ID/g

L1(-) for 64 NOT A- PD1 at 1 hour pi 4.2 (±0.8) %ID/g for L1(+) and 3.5 (±1.7) %ID/g for L1(-) for 64 NOT PD1 at 1 hour pi 2.7 (±1.1) %ID/g for L1(+) and 0.8 (±0.4) %ID/g for L1(-) for 64 NOT PD1 at 1 hour pi

Yes, spleen 4.0 (±3.1) %ID/g, 5.5 (±1.4) %ID/g and 1.4 (±0.4) %ID/g for 64 DOT A- PD1, 64 NOT PD1, and 64 Cu-NOT PD1 respectively ( 18 )

68Ga- PD-1 ectodomains (DOT

A-/ NOT HAC, aglycosylated DOT A-/NOT A-HACA) Engineer ed anti-human Not specified

NSG mice 6 to 8 weeks old Immune deficient CT26 (mouse colon cancer)

L1(+)

or

L1(-)

0.7 to 3.7 MBq (10 to 15 µg) Imaging and ex vivo biodistribution at 1

hour pi

3.8 (±1.6) %ID/g for PD- L1(+) and 1.7 (±1.3) %ID/g for

L1(-) for 68 NOT PD1 at 1 hour pi 2.8 (±1.5) %ID/g for L1(+) and 0.8 (±0.1) %ID/g for L1(-) for 68 DOT PD1 at 1 hour pi

Yes, spleen 3.5 (±0.6) %ID/g and 0.2 (±0.2) %ID/g for 68

NOT PD1 and 68 DOT A- PD1 respectively PET 64 FN3 L1

Small molecule

human No L1 3.7 (±0.4) MBq (8 to 10 µg) Imaging at 0.5, 1, 4, 18, and 24 hours pi followed by ex vivo biodistribution

5.6 (±0.9) %ID/g at 24 hours pi for CT26/

L1 tumors No ( 19 ) MB-231 (human br east cancer)

3.6 (±0.5) %ID/g at 24 hours pi for MDA- MB-231 tumors

PET 68 WL12 L1 binding peptide) Engineer ed anti-human No

NSG mice 6 to 8 weeks old Immune deficient

Human cell lines

±7.4

MBq for imaging

and ±0.9

MBq for ex vivo

biodistribution

Imaging and ex vivo biodistribution at 15, 60, and 120

min pi

11.56 (±3.18) %ID/g, 4.97 (±0.8) %ID/g and 1.9 (±0.1) %ID/g for

L1,

MB-231

and SUM149 tumors respectively at 60

min pi No ( 20 ) PET 64 WL12 L1 binding peptide) Engineer ed anti-human No

NSG mice 5 to 6 weeks old Immune deficient Human cell lines: H226, HCC827,

L1+, hPDL1-, MDAMB231 ±7.4 MBq for imaging and ±0.74 MBq for ex vivo biodistribution

Imaging and ex vivo biodistribution at 120

min

pi Treatment with atezolizumab

24

hours

prior to tracer injection (20

mg/kg)

~5.5 %ID/g,~8 %ID/g,~18 %ID/g,~5 %ID/g,~8 %ID/g for H226, HCC827, CHO- PDL1+,

PDL1- and MDAMB231 r espectively at 120 min pi Tr eatment r educed uptake

in all cell lines? T

umors

models?

Yes, spleen ~4 %ID/g, after tr

eatment ~3.5 %ID/g ( 21 ) PD1 - antibodies PET 64 PD-1 antibody

Hamster anti- mouse

No

Tr

eg+transgenic mice

(Foxp3+.LuciDTR) Immune competent B16F10 (mouse melanoma) 7.4 (±0.4) MBq (10– 12 µg) Blocking with fivefold molar excess Imaging and ex vivo biodistribution at 1

hour

,

24

hours, and 48

hours pi

7.4 (±0.71) %ID/g for

block vs 4.51 (±0.26)

%ID/g for blocking 48 hours

pi

Yes, spleen 23.04 (±4.97) %ID/g for non- block vs 14.39±0.53) %ID/g for blocking 48 hours

pi ( 22 ) Table 1 Continued Continued copyright.

on November 18, 2020 at University of Groningen. Protected by

(6)

Type of Imaging

Tracer

Origin and reactivity

Cr oss reactivity Animal model Tumor model Tracer dose

Imaging / biodistribution time point

Tumor uptake

Uptake lymphoid tissue

Ref PET 89Zr - pembr olizumab

Humanized anti- human

Not specified

NSG and humanized NSG mice (hNSG) A375 (human melanoma) 3.2 (±0.4) MBq (15 to 16 µg) Imaging at 1, 4, 18, 24, 48, 72, 96, 120 and 144

hours

pi, ex vivo biodistribution at 144

hours pi

1.8 (±0.4) %ID/g for NSG and 3.2 (±0.7) %ID/g for hNSG at 144

hours pi

Yes, spleen ~19 %ID/g for NSG and ~28 %ID/g for hNSG at 144

hours pi ( 23 ) 64Cu- pembr olizumab 7.4 (±0.4) MBq (20 to 25 µg) Imaging at 1, 4, 18, 24 and 48 hours pi, ex

vivo biodistribution at 48 hours

pi

5.7 (±0.6) %ID/g for NSG, 9.4 (±2.5) %ID/g for hNSG and 5.9 (±2.1) %ID/g for hNSG block at 48

hours pi

Yes, spleen ~6.5 %ID/g for NSG,~10.5 for hNSG, and ~7%

ID/g for hNSG block at 48 hours pi PET 89Zr - pembr olizumab

Humanized anti- human

No

ICR (CD-1) mice and Hsd

Dawley

rats, 5 weeks old Immune competent

No tumor model Mice: 5 to 10 MBq (7 to 14 µg) Rats: 50 MBq (14 µg) Imaging at 3, 6, 12, 24, 48, 72, and 168 hours pi, ex vivo biodistribution at 168 hours pi No tumor model

Yes, spleen ~2.5 %ID/g for mice and ~1%

ID/g for rats 168

hours pi

(

24

)

NSG mice and humanized NSG mice engrafted with human PBMCs

SCID),

5–8 weeks old

No tumor model; PBMC engraftment

No tumor model

Yes, spleen ~8 %ID/g for NSG and ~4.5 %ID/g for

SCID) at 168 hours pi PET 89Zr - nivolumab

Humanized anti- human

No

NSG mice and humanized NSG mice engrafted with human PBMCs

SCID

3–5 weeks old

A549 (human lung cancer)

5 to 10 MBq (7 to 14

µg)

Imaging at 3, 6, 12, 24, 48, 72, and 168

hours pi,

and ex vivo biodistribution at 168

hours pi.

3.88 (±0.38) %ID/g for NSG and 9.85 (±2.73) %ID/g for

SCID at

168

hours

pi

2.85 (±0.39) %ID/g for

PBL IgG contr

ol at

168

hours

pi

Yes, 7.48 (±0.47) %ID/g for NSG and 4.32 (±0.40) %ID/g for

SCID) at 168

hours

pi 3.05 (±0.79) %ID/g for

SCID IgG contr ol at 168 hours pi ( 25 ) PET 89Zr - nivolumab

Humanized anti- human

Af finity for cynomolgus monkey Healthy human primates – 54.5 (±11.0) MBq (237 µg) Imaging at 24 hours, 96 hours, 144 hours and 192 hours – Yes, spleen at 192 hours SUV=17.63 Blocking 1 mg/kg at 192 hours SUV=2.5, 3 mg/kg SUV=2.62 ( 26 ) PET 64Cu- pembr olizumab

Humanized anti- human

No

Humanized NSG mice

293T (human embryonic kidney cell line) expr

essing

L1

7.4 (±0.4) MBq (20 to 25 µg) Dynamic PET scans on 1, 2, and 4

hour pi during 3 min, at 18 and 24 hours pi during 5 min, at 24 hours pi during 10

min and at and

48

hours pi during 15

min

Ex vivo biodistribution at 1, 12, 24, and 48

hours pi

14.8 (±1.2) %ID/g for 293T tumors at 48

hours pi

0.44 (±0.01) %ID/g for A375 tumors at 48

hours pi

Yes, spleen (17.5%±1.6 %ID/g) at 48

hours

pi

(

27

)

A375 (human melanoma)

PDL1 + PD1 antibodies PET 64 PD-1 and 64 L1 antibody

Murine anti- mouse

No C57BL/6N mice deficient mice deficient mice Immune competent B16F10 (mouse melanoma) 1.13 (±0.31) MBq (1.5 µg) 64 PD-1 and 6.38 (±0.35) MBq (20 µg) 64 L1

Dynamic PET scan during 45–55 and 15–20

min at 24 hours pi for 64 Cu-PD-1 and 64 L1

respectively Ex vivo biodistribution at 48 hours

pi ±14 %IA/cm 3 in B16F10 tumor at 24 hours pi in vivo for 64 PD-1 and 64 L1 ±12 %IA/cm 3 in B16F10 tumor at 24 hours pi ex vivo for 64 L1

Yes, spleen (±20 %IA/cm

3) and lymph nodes

(20%–30%IA/cm

3) for 64

PD-1, spleen (15

%IA/cm

3), lymph nodes (7.5%–15%IA/cm 3) and BA T (±12 %IA/cm 3) for 64 L1

Detection of PD-1 +TILs after

py

L1 upr

egulation (mainly in lung) by

IFN-γ tr eatment visualized ( 28 ) WT ; type; AlF , aluminum fluoride; BA T, br

own adipose tissue; DOT

A,

tetraazacyclododecane- N,N',N

″,N'

″-tetraacetic acid ; DTP

A, diethylenetriaminepentaacetic acid; EGFR, epidermal gr

owth factor r

eceptor; %ID/g, per

centage of injected dose per gram;

IFN-γ, interfer

gamma; KO,

out; LPS,

lipopolysaccharide; NOT

A,

N,N',N''-triacetic acid; NSCLC,

small cell lung cancer; NSG, NOD SCID gamma; PBMC, peripheral blood mononuclear cell; PD-1, pr

ogrammed cell death pr

otein 1;

L1, pr

ogrammed

ligand 1; PDX,

derived xenograft; PET

, positr

on emission

tomography; pi,

injection; SPECT

, single photon emission CT

; TILs, tumor

- infiltrating lymphocytes.

Table 1

Continued

copyright.

on November 18, 2020 at University of Groningen. Protected by

(7)

antibody (mAb), not cross- reactive with murine PD-1, we aimed to study the biodistribution of zirconium-89 (89Zr)

radiolabeled pembrolizumab in melanoma- bearing humanized NOG mice (huNOG) engrafted with HSCs using positron- emission tomography (PET) imaging. To enable consecutive clinical translation of this approach, we developed and validated a good manufacturing prac-tices (GMP) compliant production process for 89Zr-

pem-brolizumab. Finally, we put our data in perspective by summarizing results from current in vivo preclinical studies with PD-1 and PD- L1 targeting radiolabeled molecules.

METHODS Cell lines

The human melanoma cell line A375M was purchased from the American Type Culture Collection. Cell lines were confirmed to be negative for microbial contamina-tion and were authenticated on August 6, 2018, by Base-Clear using short tandem repeat profiling. A375M cells were routinely cultured in Roswell Park Memorial Insti-tute 1640 medium (Invitrogen) containing 10% fetal calf serum (Bodinco BV), under humidified conditions at 37°C with 5% CO2. Cells were passaged 1:10, twice a week. For in vivo experiments, cells in the exponential growth phase were used.

Development of 89Zr-pembrolizumab and 89Zr-IgG

4

First, the buffer of pembrolizumab (25 mg/mL, Merck) was exchanged for NaCl 0.9% (Braun) using a Vivaspin-2 concentrator (30 kDa) with a polyethersulfon filter (Sartorius). Next, pembrolizumab was conjugated with the tetrafluorphenol- N- succinyldesferal- Fe(III) ester (TFP- N- sucDf; ABX) as described earlier, in a 1:2 TFP- N- sucDf:mAb ratio.30 Conjugated product was purified from

unbound chelator using Vivaspin-2 concentrators and stored at −80 °C. On the day of tracer injection, N- sucDf- pembrolizumab was radiolabeled with 89Zr, delivered

as 89Zr- oxalate dissolved in oxalic acid (PerkinElmer),

as described previously.30 For in vivo studies,

pembroli-zumab was radiolabeled at a specific activity of 250 MBq/ mg. IgG4 control molecule (Sigma- Aldrich) was conju-gated with TFP- N- sucDf at a 1:3 molar ratio, followed by radiolabeling with 89Zr at similar specific activity of 250

MBq/mg.

Quality control of 89Zr-pembrolizumab

Size exclusion high- performance liquid chromatography (SE- HPLC) was used to determine the final number of TFP- N- sucDf ligands per antibody (chelation ratio). SE- HPLC analysis was also performed to assess potential aggrega-tion and fragmentaaggrega-tion for both N- sucDf- pembrolizumab and 89Zr- pembrolizumab. An HPLC system (Waters)

equipped with an isocratic pump (Waters), a dual wave-length absorbance detector (Waters), in- line radioactivity detector (Berthold) and a TSK- GEL G3000SWXL column (Tosoh Biosciences) was used with phosphate buffered

saline (PBS, sodium chloride 140.0 mmol/L, sodium hydrogen phosphate 0.9 mmol/L, sodium dihydrogen phosphate 1.3 mmol/L; pH 7.4) as mobile phase (flow 0.7 mL/min). Radiochemical purity of 89Zr-

pembroli-zumab was measured by trichloroacetic acid precipita-tion assay.31 Immunoreactivity of 89Zr- pembrolizumab was

analyzed by a competition binding assay with unlabeled pembrolizumab. Nunc- immuno break apart 96- wells plates (Thermo Scientific) were coated overnight at 4°C with 100 µL of 1 µg/mL PD-1 extracellular domain (R&D Systems) in PBS, set to pH 9.6 with Na2CO3 2M. Plates were washed with 0.1% Tween 80 in PBS and blocked for 1 hour at room temperature (RT) with 150 µL 1% human serum albumin (Albuman, Sanquin) in PBS. Multiple 1:1 mixtures of 89Zr- pembrolizumab with unlabeled

pembroli-zumab were prepared, using a fixed concentration of

89Zr- pembrolizumab (7000 ng/mL) and varying

concen-trations of unlabeled pembrolizumab (from 3.75 ng/mL to 12.5×106 ng/mL). Of each mixture, 100 µL was added

to the 96- wells plate and incubated for 2 hours at RT. After washing twice with washing buffer, radioactivity in each well was counted using a gamma counter (Wizard2 2480–

0019, SW 2.1, PerkinElmer). Counts were plotted against the concentration of competing unlabeled pembroli-zumab. The half maximal inhibitory concentration (IC50) was calculated using GraphPad Prism 7 (GraphPad soft-ware). Immunoreactivity was expressed as the IC50 value divided by the 89Zr- pembrolizumab concentration to

calculate the immune reactive fraction (IRF).

Animal studies

All animal studies were approved by the Institutional Animal Care and Use Committee of the University of Groningen. Studies were performed in humanized NOG mice (NOD.Cg- Prkdcscid Il2rgtm1Sug/JicTac, Taconic) and

non- humanized NOG mice (Taconic) were used for control experiments. HuNOG mice are sublethally irra-diated 3 weeks after birth and subsequently reconstituted with human CD34+ hematopoietic stem cells derived from

fetal cord blood to express a functional human immune system including B- cells, T- cells, NK- cells, dendritic cells and monocytes. HuNOG and NOG mice were subcutane-ously xenografted with 5×106 A375M human melanoma

cells in 300 µL of a 1:1 mixture of PBS and Matrigel (BD Biosciences) on the right flank. Tumor growth was assessed by caliper measurements. When tumor volumes reached 100 to 200 mm3 (after 2 weeks), 2.5 MBq 89Zr-

pembroli-zumab (10 µg) was administered via retro- orbital injec-tion. Mice were anesthetized using isoflurane/medical air inhalation (5% induction, 2.5% maintenance).

The first group of huNOG mice received 10 µg

89Zr- pembrolizumab (n=5). In addition, a second group

of huNOG mice xenografted with the same tumor model received a co- injection of 10 µg 89Zr- pembrolizumab and

90 µg unlabeled pembrolizumab (n=4). To a third group of huNOG mice, 2.5 MBq 89Zr- IgG

4 control (10 µg) was administered (n=4). Control NOG mice received 10 µg

89Zr- pembrolizumab (n=4).

copyright.

on November 18, 2020 at University of Groningen. Protected by

(8)

PET imaging and ex vivo biodistribution

On day 7 post tracer injection (pi), PET scanning was performed. We selected this day based on optimal tumor- to- blood ratio and technical aspects, including feasible tracer specific activity and animal welfare. Mice were placed in a Focus 220 rodent scanner (CTI Siemens) on heating matrasses. Acquisition time was 60 min. A trans-mission scan of 515 s was performed using a 57Co point

source to correct for tissue attenuation. After scanning, mice were sacrificed for ex vivo biodistribution. Bone marrow was collected from the femur bone by centrifugal- based separation. All other organs were dissected and counted in a gamma- counter (Wizard2 2480–0019, SW 2.1,

PerkinElmer). Tracer uptake in each organ was expressed as percentage of the injected dose per gram tissue weight, calculated by the following formula: %ID/g = (activity in tissue (MBq)/total injected activity (MBq))/tissue weight (g)×100. To compare ex vivo and in vivo uptake, ex vivo uptake was also calculated as mean radioactivity per gram tissue, adjusted for total body weight (SUVmeanex vivo), calculated with the following formula: SUVmeanexvivo = (activity in tissue (MBq)/total injected activity (MBq))×-mouse weight (g). Calculations are corrected for decay and background.

PET data was reconstructed and in vivo quantification was performed using PMOD software (V.4.0, PMOD tech-nologies LCC). Three- dimensional regions of interest were drawn around the tumor. For other organs and tissues, a size- fixed sphere was drawn in representative tissue parts. PET data was presented as mean standardized uptake value (SUVmean in vivo), calculated by the following formula: SUVmean (g/mL) = (activity concentration (Bq/ mL)/applied dose (Bq))×weight (kg)×1000.

Autoradiography

Tumor and spleen from ex vivo biodistribution studies were formalin- fixed and paraffin embedded (FFPE). FFPE tissue blocks where cut into slices of 4 µM. These slices were exposed to a phosphor imaging screen (Perki-nElmer) for 72 hours and then scanned using a Cyclone phosphor imager (PerkinElmer).

Immunohistochemistry

Subsequent slices of the same tumor, spleen and mesen-teric lymph node tissue were stained for H&E, CD3, CD8 and PD-1. FFPE tumor, spleen and lymph node tissue were cut into 4 µm slices using a microtome (Microm Hm 355 s, Thermo Scientific) and mounted on glass slides. Tissue sections were deparaffinized and rehydrated using xylene and ethanol. Heat- induced antigen retrieval was performed in citrate buffer (pH=6) at 100°C for 15 min. Endogenous peroxidase was blocked by 30 min incuba-tion with 0.3% H2O2 in PBS. For CD3 staining, slides were incubated with rabbit anti- human CD3- antibody (Spring bioscience; clone SP162) in a 1:100 dilution in PBS/1% bovine serum albumin (BSA) at RT for 15 min. For CD8 staining, slides were incubated with rabbit anti- human CD8- antibody (Abcam; clone SP16) in a 1:50 dilution in

PBS/1% BSA at 4°C overnight. For PD-1 staining, slides were incubated with rabbit anti- human PD-1- antibody (Abcam, clone EPR4877(2)) in a 1:500 dilution in PBS/1% BSA at RT for 30 min. Human tonsil or lymph nodes tissues sections served ad positive control and were incubated with either CD3, CD8 or PD-1 antibody. As a negative control human tonsil or lymph nodes sections were incubated with rabbit IgG monoclonal antibody (Abcam, clone EPR25A) or PBS/1% BSA.

For CD3, CD8 and PD-1 staining, incubation with secondary antibody (anti- rabbit EnVision+, Dako) was

performed for 30 min, followed by application of diam-inobenzidine chromogen for 10 min. Hematoxylin counterstaining was applied and tissue sections were dehydrated using ethanol and imbedded using mounting medium (Eukitt). H&E staining served to analyze tissue viability and morphology. Digital scans were acquired by a Nanozoomer 2.0- HT multi slide scanner (Hamamatsu).

89Zr-pembrolizumab manufacturing according to GMP

To enable clinical application, GMP- compliant 89Zr-

pem-brolizumab was developed. First, N- sucDf- pempem-brolizumab intermediate product was produced on a larger scale (60 mg batch, divided in 2.5 mg aliquots) and subse-quently radiolabeled with 89Zr, followed by purification,

dilution and sterile filtration (online supplemental figure S1). Release specifications were defined, as shown in online supplemental table S1. All analytical methods for quality control (QC) were validated. According to protocol validation of both N- sucDf- pembrolizumab and

89Zr- pembrolizumab, manufacturing consisted of three

independent validation runs, including complete release QC. Stability of N- sucDf- pembrolizumab stored at −80 °C was studied up to 6 months and stability of 89Zr-

pembroli-zumab was determined up to 168 hours at 2°C to 8°C stored in a sterile, type 1 glass injection vial. In addition, in use stability was demonstrated at RT in a polypropylene syringe for up to 4 hours (online supplemental table S2).

Statistical analysis

Data are presented as median±IQR. A Mann- Whitney U test, followed by a Bonferroni correction was performed to compare groups (GraphPad, Prism 7). P values ≤0.05 were considered significant. If not indicated otherwise, results were not statistically significant.

RESULTS

89Zr-pembrolizumab development for in vivo studies

We optimized the conjugation processes of pembroli-zumab with the TFP- N- sucDf chelator and its subsequent radiolabeling with 89Zr. For in vivo studies, N- sucDf-

pembrolizumab was produced with >60% yield and average 1.7 chelators per antibody (online supplemental figure S2, table S1). N- sucDf- pembrolizumab was subsequently radiolabeled with 89Zr at a specific activity of 250 MBq/

mg, with radiochemical purity of >95% after purification. Both N- sucDf- pembrolizumab and 89Zr- pembrolizumab

copyright.

on November 18, 2020 at University of Groningen. Protected by

(9)

were stable, as shown in online supplemental table S1, S2 and figure S2. Immunoreactivity was not impaired by conjugation or radiolabeling.

89Zr-pembrolizumab imaging and biodistribution in humanized

mice

PET imaging revealed 89Zr- pembrolizumab uptake in

tumor, but also in healthy tissues, including liver, spleen and lymph nodes, of A375M tumor- bearing huNOG mice (figure 1A,B). Consistent with these results, ex vivo biodis-tribution at day 7 pi showed highest 89Zr- pembrolizumab

uptake in spleen (SUVmean 30.5, IQR 15.8 to 67.7), mesen-teric lymph nodes (SUVmean 20.4, IQR 8.0 to 25.2), bone marrow (SUVmean 14.5, IQR 6.1 to 32.8), thymus (SUVmean 1.3, IQR 1.1 to 2.1), liver (SUVmean, IQR 6.0, IQR 3.4 to 9.9) and tumor (SUVmean 5.1, IQR 3.3 to 8.9) (figure 1C, online supplemental table S3).

Tumor uptake of 89Zr- pembrolizumab was variable and

slightly higher than tumor uptake observed for 89Zr- IgG 4 control, however not significant due to small groups of mice (SUVmean 5.1, IQR 3.3 to 8.9 vs SUVmean 3.5, IQR 2.7 to 4.4) (figure 1C). This may be explained by low PD-1 expression found in all tumors by immunohistochemical

(IHC) analysis (figure 2). 89Zr- pembrolizumab tumor-

to- blood ratio also did not differ from 89Zr- IgG

4 control (figure 1D).

89Zr- pembrolizumab in huNOG mice showed higher

uptake in lymphoid tissues compared with 89Zr- IgG 4 control: spleen (SUVmean 13.9, IQR 7.1 to 21.4, NS, p=0.254), mesenteric lymph nodes (SUVmean 2.3, IQR 1.4 to 4.4, NS, p=0.114), salivary gland (SUVmean 2.1, IQR 1.2 to 2.9, NS, p=0.635), bone marrow (SUVmean 8.8, IQR 7.6 to 10.0, NS, p=1.714) and thymus (SUVmean 0.5, IQR 0.4 to 1.1, p=0.1714), indicating that 89Zr- pembrolizumab

uptake in these tissues is, at least partly, PD-1- mediated.

89Zr- pembrolizumab tissue- to- blood (T:B) and tissue- to-

muscle (T:M) ratios in lymphoid organs confirmed high uptake in these tissues (figure 1D,E). Additionally, rela-tively high 89Zr- IgG

4 uptake was found in spleen, bone marrow and liver compared with other organs, suggesting

89Zr- pembrolizumab uptake in these tissues is also due

to Fcγ receptor (FcγR)- binding of the antibody’s Fc- tail. High 89Zr- IgG4 uptake was less evident in lymph nodes and thymus.

Figure 1 In vivo PET imaging and ex vivo biodistribution of 89Zr- pembrolizumab in immunocompetent humanized NOG

mice. Mice were xenografted with A375M tumor cells and received tracer injection at day 0. For blocking studies huNOG mice received a 10- fold excess of unlabeled pembrolizumab (huNOG excess). As a control for non- specific uptake huNOG mice were injected with 89Zr- IgG

4. PET imaging performed on day 7 post injection (pi). (A) In vivo PET examples (maximum intensity

projections) at day 7 pi showing uptake in tumor (T), axillary lymph nodes (LN), liver (L) and spleen (S). (B) In vivo uptake of

89Zr- pembrolizumab in spleen, lymph nodes (axillary), liver and tumor, at day 7 pi. Uptake is expressed as SUV

mean. (C) Ex vivo

biodistribution of 89Zr- pembrolizumab in humanized NOG mice. Uptake is expressed as mean radioactivity per gram tissue,

adjusted for total body weight (SUVmeanex vivo). Data expressed as median±IQR *p≤0.05. BAT, brown adipose tissue; huNOG, humanized NOG mice; MLN, mesenteric lymph nodes; PET, positron emission tomography.

copyright.

on November 18, 2020 at University of Groningen. Protected by

(10)

89Zr- pembrolizumab spleen uptake in huNOG mice

was blocked by the addition of a 10- fold excess unlabeled pembrolizumab (SUVmean 30.5, IQR 15.8 to 67.7 versus SUVmean 5.1, IQR 4.3 to 7.0, p=0.032) (figure 1B,C). Uptake in other lymphoid organs and liver was also reduced by addition of unlabeled mAb dose, whereas uptake in non- lymphoid tissues was unaffected (online supplemental table S3). Tracer activity in blood pool was increased by addition of unlabeled mAb (SUVmean 0.1, IQR 0.0 to 1.8 to SUVmean 2.2, IQR 1.4 to 7.4), but uptake in tumor did not change.

Autoradiography confirmed PET imaging results on a macroscopic level, showing high uptake in spleens of huNOG mice compared with spleens of mice that received an additional unlabeled pembrolizumab dose (figure 3). Furthermore, comparable tumor uptake was found for different dose groups. IHC analysis on spleen and lymph node tissue of huNOG mice revealed that PD-1, CD3 and CD8 positive cells were present. CD3 and CD8 cells were also present in tumor tissue of huNOG mice (figure 2), however, PD-1 staining of these tumors was negative.

89Zr- pembrolizumab biodistribution in NOG control

mice clearly showed a different pattern than in huNOG mice, with high uptake in liver (SUVmean 16.9, IQR 5.1 to 26.2) and spleen (SUVmean 49.6, IQR 16.6 to 135.6),

whereas 89Zr- pembrolizumab tumor uptake in NOG mice

was similar to huNOG mice (SUVmean 9.3, IQR 4.5 to 15.7 vs SUVmean 5.1, IQR 3.3 to 8.9) (online supplemental figure S3). High 89Zr- pembrolizumab spleen uptake

in this model may be unexpected, since limited T- cells are present in NOG mice (online supplemental figure

Figure 2 IHC analysis of spleen, mesenteric lymph node and tumor tissue humanized NOG mice. Formalin- fixed and paraffin

embedded tissue blocks where cut into slices of 4 µM and stained for PD-1, CD3 and CD8 (40x). H&E staining served to analyze tissue viability and morphology (40x). Scalebar: 50 µm. IHC, immunohistochemical; PD-1, programmed cell death protein-1.

Figure 3 Autoradiography of spleen and tumor tissue

humanized NOG mice (huNOG). Formalin- fixed and paraffin embedded tissue blocks where cut into slices of 4 µM. These slices were exposed to a phosphor imaging screen for 72 hours and were then scanned using a Cyclone phosphor imager.

copyright.

on November 18, 2020 at University of Groningen. Protected by

(11)

S3). However, high spleen uptake in severely immuno-compromised mice has been described previously and is potentially Fcγ receptor- mediated.23 24 32 Moreover,

spleen weights in NOG mice were lower than in huNOG mice (NOG: 0.017 g±0.015 g; huNOG: 0.037 g±0.016 g, p=0.036), which resulted in higher tracer uptake expressed as %ID per gram spleen tissue for NOG mice. A low spleen weight may result from high radiosensitivity of NOG splenocytes, which can lead to toxicity.33

Critical steps in 89Zr-pembrolizumab manufacturing

The production processes for N- sucDf- pembrolizumab intermediate product and 89Zr- pembrolizumab for in vivo

studies were modified to comply with GMP requirements. In the conjugation reaction, pH is increased from 4.5 to 8.5, performed in small titration steps, as described earlier by Verel et al.30 During this pH transition, precipitation

occurred at 6.5 to 7.0, which was re- dissolved at pH >7.5. No precipitation was observed when pH was changed abruptly, for example, by buffer exchange, to pH 8.5 during conjugation and to pH 4.5 for removal of Fe(III). This indicates potential instability of pembrolizumab at pH 6.5 to 7.0. Formation of aggregates may be explained by the fact that pembrolizumab is an IgG4 type mAb, which forms non- classical disulfide bonds. In contrast, IgG1 type antibodies can only form classical disulfide bonds. There are many other determinants of antibody stability besides disulfide bond formation, however, this phenomenon was not seen previously with the radiolabeling of IgG1 type antibodies.31 33 34

Immunoreactivity was not affected when pembroli-zumab showed precipitation during pH transition, demonstrated by comparable IRF for precipitated N- sucDf- pembrolizumab and for non- precipitated N- sucDf- pembrolizumab (online supplemental figure S4). However, it is unknown whether the pembrolizumab struc-ture is modified by the formation of precipitates. There-fore, the method for pH transition by buffer exchange was incorporated in the conjugation protocol for pembroli-zumab. Production of clinical grade 89Zr- pembrolizumab

was performed as previously described by Verel et al.30

89Zr-pembrolizumab GMP validation

Three consecutive batches of conjugated and radiola-beled pembrolizumab were produced at clinical scale and complied with all release specifications (online supplemental tables S1 and S2), indicating that our process for manufacturing clinical grade 89Zr-

pembroli-zumab is consistent and robust. 89Zr- pembrolizumab

was obtained with a specific activity of 37 MBq/mg and mean IRF of 1.35±0.6 (n=3). Stability studies revealed that N- sucDf- pembrolizumab remained compliant to release specifications up to 6 months storage at −80°C, therefore N- sucDf- pembrolizumab shelf- life was set at 6 months. Stability studies are ongoing and shelf- life may be extended if future time points remain within spec-ifications. Data obtained during process development and validation were used to compile the investigational

medicinal product dossier (IMPD), which includes all information regarding quality control, production and validation of 89Zr- pembrolizumab. Based on this IMPD, 89Zr- pembrolizumab has been approved by competent

authorities for use in clinical studies.

DISCUSSION

This study reveals 89Zr- pembrolizumab whole- body

distri-bution in tumor- bearing huNOG mice established with a broad set of developed immune cells. Tumor uptake of

89Zr- pembrolizumab was markedly lower than uptake in

lymphoid tissues such as spleen, lymph nodes and bone marrow, but higher than uptake in other organs. Impor-tantly, high uptake in lymphoid tissues could be reduced with a 10- fold excess of unlabeled pembrolizumab. This contrasts with 89Zr- pembrolizumab tumor uptake,

which was not reduced by the addition of unlabeled pembrolizumab.

Our study nicely shows the in vivo behavior of 89Zr-

pem-brolizumab, which, apart from IgG pharmacokinetics determined by its molecular weight and Fc tail, is predom-inantly driven by its affinity for PD-1 (Kd:~30 pM). The PD-1 cell surface receptor is primarily expressed on acti-vated T- cells and pro B- lymphocytes, which are abun-dantly present in our huNOG mouse model. Lymphocytes are highly concentrated in organs that are key players of the immune system: lymph nodes, spleen, thymus, bone marrow as well as tonsils, adenoid and Peyer’s patches. From our PET imaging and ex vivo biodistribution data, we learned that 89Zr- pembrolizumab distributed mainly

to lymphoid organs, where PD-1 expressing immune cells are present.

89Zr- pembrolizumab showed relatively low and variable

tumor uptake, however, this uptake could be visualized with PET imaging 7 days pi and was higher than in non- lymphoid tissues. We hypothesized there may be PD-1- mediated 89Zr- pembrolizumab tumor uptake, but we also found tumor uptake for 89Zr- IgG4, suggesting part of the 89Zr- pembrolizumab tumor uptake is FcγR- mediated. In

our mouse model, few PD-1 positive immune cells may have traveled to the tumor, thereby potentially limiting

89Zr- pembrolizumab tumor uptake. Interestingly, the

addition of unlabeled pembrolizumab did not influence tumor uptake. This is likely caused by substantial increase of 89Zr- pembrolizumab in blood pool as a direct

conse-quence of adding excess unlabeled pembrolizumab, warranting a continuous pembrolizumab supply to the tumor.

Ex vivo immunohistochemical analysis revealed CD3 and CD8 positive lymphocytes were present in tumor, but limited PD-1- expression was found. Immune checkpoint protein expression status in tumor- infiltrating lympho-cytes is highly dynamic.35 36 This so- called ‘immune

phenotype’ depends on several factors, including tumor type, location and mutational burden. Our results indi-cate that, whereas PD-1 expression may demonstrate large

copyright.

on November 18, 2020 at University of Groningen. Protected by

(12)

variation, 89Zr- pembrolizumab PET imaging is able to

capture PD-1 dynamics in both tumor and healthy tissues. Compared with earlier preclinical studies with radio-labeled pembrolizumab in the hNSG model, we found higher 89Zr- pembrolizumab uptake in spleen and other

lymphoid tissues.23 24 This likely reflects the presence of

multiple hematopoietic cell lineages, including B- cells, T- cells, NK- cells, dendritic cells and monocytes, and thus higher PD-1 expression, in our huNOG model compared with the hNSG model. Molecular imaging studies with radiolabeled antibodies generally show distribution to the spleen. It also known that Fc/FcγR- mediated immu-nobiology of the experimental mouse model plays a key role in the in vivo biodistribution and tumor targeting.33

In our mouse model, we also observed 89Zr- IgG

4 uptake in lymphoid tissues, indicating 89Zr- pembrolizumab uptake

in these organs may have an FcγR- mediated component. For most radiolabeled antibodies without an immune target, spleen uptake in patients is ~5 %ID/kg.37 This

supports the idea that, independent of their target, anti-bodies often show distribution to the spleen. However, spleen uptake may be higher if PD-1 or PD- L1 is present.

Pembrolizumab has an IgG4κ backbone with a stabi-lizing SER228PRO sequence alteration in the Fc- region to prevent the formation of half molecules. The IgG4 backbone of pembrolizumab may slightly differ from the IgG4 control molecule that we used for our experiments, however, FcγR- binding affinity and kinetics of pembroli-zumab appears to be very similar to IgG4.38 We, therefore,

consider the used IgG4 control molecule to provide a useful indication of the extent of FcγR- mediated uptake. In this respect, FcγR- mediated uptake may be present in the spleen but potentially also in liver and tumor, since these tissues demonstrate relatively high uptake of

89Zr- IgG 4.

PD-1 is predominantly expressed on activated T- cells while its ligand PD- L1 is expressed by a broader range of immune cells as well as tumor cells. It is therefore to be expected that biodistribution of antibody tracers targeting PD- L1 may deviate from the biodistribution results that we described here for 89Zr- pembrolizumab. In table 1, we

presented an overview of preclinical imaging and biodis-tribution studies using anti- PD-1 and anti- PD- L1 tracers. Data turned out to be highly variable, mostly focused on tumor and not on the immune system, and therefore not just comparable. From our results, we increasingly realize that it is extremely important for interpretation of these type of data to know the characteristics of the anti-body (origin, cross- reactivity, Fc- backbone, target, target- affinity and dose), the animal model (mouse strain, age, immune status and tumor cell line) and time points, vari-ables we detailed in the table.

As for preclinical studies, data on the distribution of PD-1 and PD- L1 targeting antibodies to lymphoid organs in patients is still limited. A clinical imaging study in 13 patients demonstrated modest 89Zr- nivolumab spleen

uptake of SUVmean 5.8±0.7, whereas uptake of this radiola-beled antibody targeting PD-1 in other lymphoid tissues

was not addressed.39 89Zr- atezolizumab ( PD- L1

anti-body) imaging in 22 patients revealed spleen uptake with an SUVmean of 15. 89Zr- atezolizumab also distributed to

other lymphoid tissues and sites of inflammation, whereas uptake in non- lymphoid organs was low. The high spleen uptake could at least partly be explained by presence of PD- L1 in endothelial littoral cells of the spleen.40 To perceive what can be expected for 89Zr- pembrolizumab

PET imaging in patients, how results may be interpreted and potentially translated to predicting response, knowl-edge on which immune cells express PD-1 and where these cells are located in the human body is of utmost importance.

With our study, we validated the use of 89Zr-

pembroli-zumab PET imaging to evaluate PD-1- mediated uptake in tumor and immune tissues in a setting that allowed for comparing tracer uptake and whole tumor tissue analysis. To enable evaluation of 89Zr- pembrolizumab

biodistribu-tion in humans, we developed clinical grade 89Zr-

pem-brolizumab. Clinical 89Zr- pembrolizumab PET imaging

in patients with melanoma and NSCLC before treatment with pembrolizumab is currently performed at our center ( ClinicalTrials. gov Identifier NCT02760225), and may elucidate if tracer tumor uptake correlates to response and if uptake in healthy PD-1 expressing tissues correlates to toxicity.

CONCLUSION

We demonstrated the in vivo biodistribution of 89Zr-

pem-brolizumab in humanized mice, and found uptake in tumor with the highest uptake in the lymphoid system, reflecting the presence of PD-1. Insight in the in vivo behavior and biodistribution of immune checkpoint targeting monoclonal antibodies might aid in better understanding immune checkpoint inhibition therapy and could potentially help explaining variation in response as well as potential toxicity due to uptake in healthy (immune) tissues.

Twitter Elisabeth G E De Vries @VriesElisabeth

Contributors ELvdV was involved in project design and conceptualization, was involved in tracer development and GMP validation, wrote the IMPD, performed animal studies, performed ex vivo analyses, data analysis and wrote the manuscript; DG was involved in study conceptualization, data analysis, performed ex vivo analyses and wrote the manuscript; LPdJ was involved in tracer development and GMP validation, performed animal studies, performed ex vivo analyses and edited the manuscript; AJS was involved in GMP validation, wrote the IMPD and edited the manuscript; EGEdV was involved in project design and conceptualization, supervised the study and edited the manuscript; MNLdH was involved in project design and conceptualization, supervised the study and edited the manuscript. All authors read and approved the final manuscript.

Funding The research leading to these results received funding from the Innovative Medicines Initiatives 2 Joint Undertaking under grant agreement No 116106 (TRISTAN). This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation program and EFPIA.

Competing interests EGEdV reports grants from IMI TRISTAN (GA no.116106), during the conduct of the study; consulting and advisory role for NSABP, Daiichi Sankyo, Pfizer, Sanofi, Merck, Synthon Biopharmaceuticals; grants from Amgen, Genentech, Roche, Chugai Pharma, CytomX Therapeutics, Nordic Nanovector,

copyright.

on November 18, 2020 at University of Groningen. Protected by

(13)

G1 Therapeutics, AstraZeneca, Radius Health, Bayer, all made available to the institution, outside the submitted work.

Patient consent for publication Not required.

Provenance and peer review Not commissioned; externally peer reviewed.

Data availability statement Data are available upon reasonable request. The data sets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Open access This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY- NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non- commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non- commercial. See http:// creativecommons. org/ licenses/ by- nc/ 4. 0/.

ORCID iDs

Elly L van der Veen http:// orcid. org/ 0000- 0001- 6224- 8114

Marjolijn N Lub- de Hooge http:// orcid. org/ 0000- 0002- 5390- 2791

REFERENCES

1 Postow MA, Sidlow R, Hellmann MD. Immune- Related adverse events associated with immune checkpoint blockade. N Engl J Med

2018;378:158–68.

2 Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3--potential mechanisms of action. Nat Rev Immunol 2015;15:45–56.

3 van der Veen EL, Bensch F, Glaudemans AWJM, et al. Molecular imaging to enlighten cancer immunotherapies and underlying involved processes. Cancer Treat Rev 2018;70:232–44. 4 Lamberts LE, Williams SP, Terwisscha van Scheltinga AGT, et al.

Antibody positron emission tomography imaging in anticancer drug development. J Clin Oncol 2015;33:1491–504.

5 Heskamp S, Hobo W, Molkenboer- Kuenen JDM, et al. Noninvasive imaging of tumor PD- L1 expression using radiolabeled anti- PD- L1 antibodies. Cancer Res 2015;75:2928–36.

6 Josefsson A, Nedrow JR, Park S, et al. Imaging, biodistribution, and dosimetry of radionuclide- labeled PD- L1 antibody in an immunocompetent mouse model of breast cancer. Cancer Res

2016;76:472–9.

7 Chatterjee S, Lesniak WG, Gabrielson M, et al. A humanized antibody for imaging immune checkpoint ligand PD- L1 expression in tumors. Oncotarget 2016;7:10215–27.

8 Lesniak WG, Chatterjee S, Gabrielson M, et al. PD- L1 Detection in Tumors Using [(64)Cu]Atezolizumab with PET. Bioconjug Chem

2016;27:2103–10.

9 Nedrow JR, Josefsson A, Park S, et al. Imaging of programmed cell death ligand 1: impact of protein concentration on distribution of anti- PD- L1 SPECT agents in an immunocompetent murine model of melanoma. J Nucl Med 2017;58:1560–6.

10 Kikuchi M, Clump DA, Srivastava RM, et al. Preclinical immunoPET/ CT imaging using Zr-89- labeled anti- PD- L1 monoclonal antibody for assessing radiation- induced PD- L1 upregulation in head and neck cancer and melanoma. Oncoimmunology 2017;6:e1329071. 11 Truillet C, Oh HLJ, Yeo SP, et al. Imaging PD- L1 expression with

ImmunoPET. Bioconjug Chem 2018;29:96–103.

12 Heskamp S, Wierstra PJ, Molkenboer- Kuenen JDM, et al. Pd- L1 microSPECT/CT imaging for longitudinal monitoring of PD- L1 expression in syngeneic and humanized mouse models for cancer.

Cancer Immunol Res 2019;7:150–61.

13 Chatterjee S, Lesniak WG, Miller MS, et al. Rapid PD- L1 detection in tumors with PET using a highly specific peptide. Biochem Biophys Res Commun 2017;483:258–63.

14 Gonzalez Trotter DE, Meng X, McQuade P, et al. In vivo imaging of the programmed death ligand 1 by 18F positron emission

tomography. J Nucl Med 2017;25:1852–7.

15 Broos K, Keyaerts M, Lecocq Q, et al. Non- Invasive assessment of murine PD- L1 levels in syngeneic tumor models by nuclear imaging with nanobody tracers. Oncotarget 2017;8:41932–46.

16 Maute RL, Gordon SR, Mayer AT, et al. Engineering high- affinity PD-1 variants for optimized immunotherapy and immuno- PET imaging.

Proc Natl Acad Sci U S A 2015;112:E6506–14.

17 Donnelly DJ, Smith RA, Morin P, et al. Synthesis and Biologic Evaluation of a Novel 18F- Labeled Adnectin as a PET Radioligand for

Imaging PD- L1 Expression. J Nucl Med 2018;59:529–35. 18 Mayer AT, Natarajan A, Gordon SR, et al. Practical immuno- PET

radiotracer design considerations for human immune checkpoint imaging. J Nucl Med 2017;58:538–46.

19 Natarajan A, Patel CB, Ramakrishnan S, et al. A novel engineered small protein for positron emission tomography imaging of human programmed death ligand-1: validation in mouse models and human cancer tissues. Clin Cancer Res 2019;25:1774–85.

20 De Silva RA, Kumar D, Lisok A, et al. Peptide- Based 68Ga- PET

Radiotracer for Imaging PD- L1 Expression in Cancer. Mol Pharm

2018;15:3946–52.

21 Kumar D, Lisok A, Dahmane E, et al. Peptide- Based PET quantifies target engagement of PD- L1 therapeutics. J Clin Invest

2019;129:616–30.

22 Natarajan A, Mayer AT, Xu L, et al. Novel radiotracer for immunoPET imaging of PD-1 checkpoint expression on tumor infiltrating lymphocytes. Bioconjug Chem 2015;26:2062–9.

23 Natarajan A, Mayer AT, Reeves RE, et al. Development of novel immunoPET tracers to image human PD-1 checkpoint expression on tumor- infiltrating lymphocytes in a humanized mouse model. Mol Imaging Biol 2017;19:903–14.

24 England CG, Ehlerding EB, Hernandez R, et al. Preclinical pharmacokinetics and biodistribution studies of 89Zr- labeled

pembrolizumab. J Nucl Med 2017;58:162–8.

25 England CG, Jiang D, Ehlerding EB, et al. 89Zr- labeled nivolumab

for imaging of T- cell infiltration in a humanized murine model of lung cancer. Eur J Nucl Med Mol Imaging 2018;45:110–20.

26 Cole EL, Kim J, Donnelly DJ, et al. Radiosynthesis and preclinical PET evaluation of 89Zr- nivolumab (BMS-936558) in healthy non-

human primates. Bioorg Med Chem 2017;25:5407–14. 27 Natarajan A, Patel CB, Habte F, et al. Dosimetry Prediction for

Clinical Translation of 64Cu- Pembrolizumab ImmunoPET Targeting

Human PD-1 Expression. Sci Rep 2018;8:633.

28 Hettich M, Braun F, Bartholomä MD, et al. High- Resolution PET imaging with therapeutic antibody- based PD-1/PD- L1 checkpoint tracers. Theranostics 2016;6:1629–40.

29 De La Rochere P, Guil- Luna S, Decaudin D, et al. Humanized mice for the study of immuno- oncology. Trends Immunol 2018;39:748–63. 30 Verel I, Visser GWM, Boellaard R, et al. 89Zr immuno- PET:

comprehensive procedures for the production of 89Zr- labeled monoclonal antibodies. J Nucl Med 2003;44:1271–81. 31 Nagengast WB, de Vries EG, Hospers GA, et al. In vivo VEGF

imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft. J Nucl Med 2007;48:1313–9.

32 Liu H, May K. Disulfide bond structures of IgG molecules: structural variations, chemical modifications and possible impacts to stability and biological function. MAbs 2012;4:17–23.

33 Sharma SK, Chow A, Monette S, et al. Fc- Mediated anomalous biodistribution of therapeutic antibodies in immunodeficient mouse models. Cancer Res 2018;78:1820–32.

34 Dijkers ECF, Kosterink JGW, Rademaker AP, et al. Development and characterization of clinical- grade 89Zr- trastuzumab for HER2/neu immunoPET imaging. J Nucl Med 2009;50:974–81.

35 Havel JJ, Chowell D, Chan TA. The evolving landscape of

biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer

2019;19:133–50.

36 Simon S, Labarriere N. Pd-1 expression on tumor- specific T cells: friend or foe for immunotherapy? Oncoimmunology

2017;7:e1364828.

37 Bensch F, Smeenk MM, van Es SC, et al. Comparative biodistribution analysis across four different 89Zr- monoclonal antibody tracers-

The first step towards an imaging warehouse. Theranostics

2018;8:4295–304.

38 Zhang T, Song X, Xu L, et al. The binding of an anti- PD-1 antibody to FcγRΙ has a profound impact on its biological functions. Cancer Immunol Immunother 2018;67:1079–90.

39 Niemeijer AN, Leung D, Huisman MC, et al. Whole body PD-1 and PD- L1 positron emission tomography in patients with non- small- cell lung cancer. Nat Commun 2018;9:4664.

40 Bensch F, van der Veen EL, Lub- de Hooge MN, et al. 89Zr-

atezolizumab imaging as a non- invasive approach to assess clinical response to PD- L1 blockade in cancer. Nat Med

2018;24:1852–8.

copyright.

on November 18, 2020 at University of Groningen. Protected by

Referenties

GERELATEERDE DOCUMENTEN

Under the imaging conditions used, sections from untreated control plants showed no detectable autofluorescence at specific excitation (Fig. When roots were treated

this antibiosis activity is primarily based on the production of antibiotic compounds (5), while for other Pseudomonas spp., like Pseudomonas putida WCS358, antibiosis is based

Being the first of its type, the one-day event had a number of objectives related to promoting the Intergovernmental Science- Policy Platform on Biodiversity and Ecosystem Services

(f−k) Tissue level view of fluoHA (f) and DOPG (g), DSPC (h), AmBisome (i), EndoTAG-1 (j), and Myocet (k) liposome distribution at 1 hpi in stab2 ibl2 and sibling control embryos,

A time series throughout embryonic development within the brood pouch revealed accumulation of PSNP in or on lipophilic cells in the early stages of embryonic development while

Keywords: fluorescent polystyrene particles; oral exposure; fish embryos; uptake route; dermal

Immune activation induced by checkpoint treatment was predominantly observed in the tumor- draining, but not nondraining, lymph nodes and was reflected in local accumulation of CD8 +

In this review, we discuss how particle size, shape, and rigidity affect biodistribution, cellular uptake, antigen presentation and the resulting immune response in murine